You are on page 1of 12

The n e w e ng l a n d j o u r na l of m e dic i n e

Original Article

A Recombinant Vesicular Stomatitis Virus


Ebola Vaccine
J.A. Regules, J.H. Beigel, K.M. Paolino, J. Voell, A.R. Castellano, Z. Hu, P. Muñoz,
J.E. Moon, R.C. Ruck, J.W. Bennett, P.S. Twomey, R.L. Gutiérrez, S.A. Remich,
H.R. Hack, M.L. Wisniewski, M.D. Josleyn, S.A. Kwilas, N. Van Deusen, O.T. Mbaya,
Y. Zhou, D.A. Stanley, W. Jing, K.S. Smith, M. Shi, J.E. Ledgerwood, B.S. Graham,
N.J. Sullivan, L.L. Jagodzinski, S.A. Peel, J.B. Alimonti, J.W. Hooper, P.M. Silvera,
B.K. Martin, T.P. Monath, W.J. Ramsey, C.J. Link, H.C. Lane, N.L. Michael,
R.T. Davey, Jr., and S.J. Thomas, for the rVSV∆G-ZEBOV-GP Study Group*​​

A BS T R AC T

BACKGROUND
The authors’ full names, academic de- The worst Ebola virus disease (EVD) outbreak in history has resulted in more than
grees, and affiliations are listed in the 28,000 cases and 11,000 deaths. We present the final results of two phase 1 trials
Appendix. Address reprint requests to Dr.
Regules at ­jason​.­a​.­regules​.­mil@​­mail​.­mil. of an attenuated, replication-competent, recombinant vesicular stomatitis virus
(rVSV)–based vaccine candidate designed to prevent EVD.
*
A complete list of members of the
rVSV∆G-ZEBOV-GP Study Group is pro-
METHODS
vided in the Supplementary Appendix,
available at NEJM.org. We conducted two phase 1, placebo-controlled, double-blind, dose-escalation tri-
Drs. Regules and Beigel and Drs. Davey
als of an rVSV-based vaccine candidate expressing the glycoprotein of a Zaire strain
and Thomas contributed equally to this of Ebola virus (ZEBOV). A total of 39 adults at each site (78 participants in all)
article. were consecutively enrolled into groups of 13. At each site, volunteers received one
A preliminary version of this article was of three doses of the rVSV-ZEBOV vaccine (3 million plaque-forming units [PFU],
published on April 1, 2015, at NEJM.org. 20 million PFU, or 100 million PFU) or placebo. Volunteers at one of the sites re-
N Engl J Med 2017;376:330-41. ceived a second dose at day 28. Safety and immunogenicity were assessed.
DOI: 10.1056/NEJMoa1414216
Copyright © 2017 Massachusetts Medical Society. RESULTS
The most common adverse events were injection-site pain, fatigue, myalgia, and
headache. Transient rVSV viremia was noted in all the vaccine recipients after dose 1.
The rates of adverse events and viremia were lower after the second dose than
after the first dose. By day 28, all the vaccine recipients had seroconversion as as-
sessed by an enzyme-linked immunosorbent assay (ELISA) against the glycopro-
tein of the ZEBOV-Kikwit strain. At day 28, geometric mean titers of antibodies
against ZEBOV glycoprotein were higher in the groups that received 20 million
PFU or 100 million PFU than in the group that received 3 million PFU, as assessed
by ELISA and by pseudovirion neutralization assay. A second dose at 28 days after
dose 1 significantly increased antibody titers at day 56, but the effect was dimin-
ished at 6 months.
CONCLUSIONS
This Ebola vaccine candidate elicited anti-Ebola antibody responses. After vaccina-
tion, rVSV viremia occurred frequently but was transient. These results support
further evaluation of the vaccine dose of 20 million PFU for preexposure prophy-
laxis and suggest that a second dose may boost antibody responses. (Funded by
the National Institutes of Health and others; rVSV∆G-ZEBOV-GP ClinicalTrials.gov
numbers, NCT02269423 and NCT02280408.)

330 n engl j med 376;4 nejm.org  January 26, 2017

The New England Journal of Medicine


Downloaded from nejm.org on August 28, 2023. For personal use only. No other uses without permission.
Copyright © 2017 Massachusetts Medical Society. All rights reserved.
Recombinant Vesicular Stomatitis Virus Ebola Vaccine

T
he worst Ebola virus disease (EVD) laboratories for both trials, are presented here
outbreak in recorded history has resulted for the three vaccine dose levels (3 million
in more than 28,000 cases and 11,000 re- plaque-forming units [PFU], 20 million PFU, and
ported deaths.1 Although the primary strategy to 100 million PFU) that were under consideration
stop the transmission of Ebola remains the iden- for human use. On the basis of the data pre-
tification and isolation of contacts and the use of sented here and additional clinical and preclini-
appropriate personal protective equipment, the cal data, the rVSV-ZEBOV vaccine (at the dose of
development of a safe and efficacious vaccine 20 million PFU) was selected for inclusion in the
would provide an important public health tool. Partnership for Research on Ebola Vaccines in
Numerous Ebola virus vaccine candidates are in Liberia (PREVAIL) trial (ClinicalTrials.gov num-
preclinical development, and some have pro- ber, NCT02344407), a phase 3 efficacy study in
ceeded to human trials.2-5 Guinea,12 and the phase 3 Sierra Leone Trial to
An Ebola virus vaccine candidate based on an Introduce a Vaccine against Ebola (STRIVE,
attenuated, replication-competent, recombinant NCT02378753).
vesicular stomatitis virus (rVSV) has shown prom-
ise in preclinical studies. The vaccine candidate Me thods
(rVSV-ZEBOV) is genetically engineered to replace
the VSV glycoprotein with the glycoprotein from Vaccine
a Zaire strain of Ebola virus (ZEBOV). Vaccina- The rVSV-ZEBOV vaccine candidate is a live at-
tion induces replication of viral particles similar tenuated recombinant virus consisting of the
to VSV but expressing the ZEBOV surface glyco- VSV strain Indiana, with the glycoprotein of the
protein. ZEBOV glycoprotein is responsible for ZEBOV Kikwit 1995 strain replacing the gene for
receptor binding and membrane fusion between the VSV envelope glycoprotein. The resultant rVSV
ZEBOV and host target cells and the induction of construct contains surface ZEBOV glycoprotein
functional antibodies, including neutralizing that exhibits a narrower host-cell tropism in vitro
antibodies.6 than wild-type VSV, as well as considerable at-
Preclinical testing of rVSV-ZEBOV supports its tenuation of replication.13 A 2015 analysis esti-
potential efficacy. The rVSV-ZEBOV vaccine has mated a 3.4% nucleotide divergence (approxi-
been shown to be attenuated in normal and im- mately 1.6% amino acid divergence) between the
munocompromised nonhuman primates in safety ZEBOV Kikwit 1995 strain and a limited number
and immunogenicity studies.7,8 Multiple studies of genomic sequences for the currently circulat-
in cynomolgus macaques have shown that a sin- ing strain,14 although no conclusions regarding
gle administration of the vaccine confers a high the effect on immunogenicity can be made.
level of protection against lethal challenge.9,10 The vaccine was developed by the Public
Various methods of vaccine delivery (oral, intra- Health Agency of Canada, licensed to BioProtec-
nasal, or intramuscular) have shown protective tion Systems (NewLink Genetics), and most re-
efficacy in animal models.11 cently sublicensed to Merck, which is responsi-
On the basis of this preclinical experience, we ble for ongoing research and development. The
conducted phase 1, double-blind, placebo-con- sponsor of the investigational new drug (IND)
trolled, dose-escalation studies of rVSV-ZEBOV application, BioProtection Systems, was involved
at two locations in the United States: the Walter in discussions of the study design and in the
Reed Army Institute of Research (WRAIR), in study monitoring and statistical analysis; it also
Silver Spring, Maryland, and the National Insti- provided the vaccine candidate. The vaccine,
tutes of Health (NIH) Clinical Center, in Bethesda, which was manufactured according to current
Maryland. Although the studies were designed Good Manufacturing Practices, was formulated
as two independent studies, the assessments and with recombinant human serum albumin and
data collections were largely harmonized. The tris(hydroxymethyl)aminomethane buffer and was
WRAIR evaluated a single-dose strategy, where- dispensed in a vial containing 100 million PFU
as the NIH evaluated a homologous, two-dose per milliliter (lot number 003 05 13). Normal
regimen administered at study days 0 and 28. saline was used as a diluent by the study phar-
Safety and humoral-immunogenicity data through macists to formulate the doses of 3 million PFU
day 180 after vaccination, generated by the same or 20 million PFU.

n engl j med 376;4 nejm.org  January 26, 2017 331


The New England Journal of Medicine
Downloaded from nejm.org on August 28, 2023. For personal use only. No other uses without permission.
Copyright © 2017 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

Volunteers and Study Design partial-thromboplastin time; and urinalysis [red-


Both trials were phase 1, double-blind, placebo- cell count and levels of protein and glucose]) at
controlled, dose-escalation trials. The trials were baseline and 7 days and 28 days after each injec-
designed to assess the safety, reactogenicity, and tion. In addition, the WRAIR site evaluated these
immunogenicity of rVSV-ZEBOV across three laboratory variables 1 day and 3 days after in-
dose levels: 3 million PFU, 20 million PFU, and jection. Grading of adverse events was based on
100 million PFU. A total of 78 healthy adult men Food and Drug Administration toxicity grading.15
and women from the Washington, D.C.–Balti- Positivity for vaccine ZEBOV-glycoprotein nucleic
more metropolitan area were recruited accord- acid sequences was assessed in plasma, saliva,
ing to protocols that were approved by the in- and urine. At the WRAIR, samples were obtained
stitutional review board at each site. Written before the injection and on days 1, 3, 7, and 14
informed consent was obtained from all the after the injection. At the NIH site, specimens
volunteers before enrollment. Exclusion criteria were obtained on days 3 and 7 after each injec-
were active involvement in clinical care of pa- tion. Further details are available in the trial
tients; substantial contact with immunocompro- protocols, available with the full text of this ar-
mised populations, children 5 years of age or ticle at NEJM.org.
younger, or animals at risk for VSV infection;
and a history of infection with filoviruses or rVSV-ZEBOV Surveillance by RT-PCR
VSV, predisposition for exposure to filoviruses A reverse-transcriptase–polymerase-chain-reaction
or VSV, or previous receipt of a filovirus vaccine (RT-PCR) assay was used to measure potential
or VSV-vectored vaccine. Pregnant or lactating rVSV virus in the plasma, saliva, and urine,
women and persons found to have the human through amplification of the Ebola Zaire glyco-
immunodeficiency virus, hepatitis B or C virus protein gene insert of the vaccine. The assay was
infection, or clinically significant medical condi- performed at the WRAIR. Details are provided
tions at screening were excluded. in the Supplementary Appendix, available at
NEJM.org.
Study Procedures
A total of 39 adults at each site were consecu- Measurement of Antibody Responses to Ebola
tively enrolled into groups of 13 each. In each Glycoprotein
group, 3 volunteers were randomly assigned in a The primary assays for antibody response were
blinded manner to receive the control (saline an enzyme-linked immunosorbent assay (ELISA)
placebo), and 10 were assigned to receive the against the homologous Zaire–Kikwit strain
rVSV-ZEBOV vaccine at a dose of 3 million PFU, glycoprotein and a pseudovirion neutralization
20 million PFU, or 100 million PFU. Each par- assay (PsVNA) against the homologous Zaire–
ticipant received a 1-ml injection in the deltoid Kikwit strain glycoprotein. These assays were
muscle; at the NIH site, a second identical dose performed at the U.S. Army Medical Research
was administered 28 days after the first. Volun- Institute of Infectious Diseases (see the Methods
teers were assessed on days 1, 3, 7, 14, and 28 section of the Supplementary Appendix). A lim-
after the first and (if applicable) second injection. ited number of samples were also tested with the
Data on solicited adverse events related to injec- use of an ELISA against the Zaire–Mayinga
tion-site and systemic reactogenicity were col- strain glycoprotein at the Vaccine Research Cen-
lected for 14 days after each injection. Data on ter of the National Institute of Allergy and Infec-
unsolicited adverse events, changes in medical tious Diseases, with the use of methods described
status, and concomitant medication use were previously,16 to allow cross-vaccine comparisons
collected for 28 days after each injection. Blood of immunogenicity.
samples were obtained for assessment of safety
and immunologic end points. All the volunteers Statistical Analysis
had safety laboratory evaluations (including a Statistical analyses were performed with the use
complete blood count with differential; measure- of R software, version 3.3.1. For each serologic
ments of serum creatinine, alanine aminotrans- variable, data were summarized by assessment
ferase, and aspartate aminotransferase levels; day and included the geometric mean titer and
determination of the prothrombin time and 95% confidence interval, the median value, and

332 n engl j med 376;4 nejm.org  January 26, 2017

The New England Journal of Medicine


Downloaded from nejm.org on August 28, 2023. For personal use only. No other uses without permission.
Copyright © 2017 Massachusetts Medical Society. All rights reserved.
Recombinant Vesicular Stomatitis Virus Ebola Vaccine

Table 1. Characteristics of the Participants at Enrollment.*

Vaccine, 3 Million PFU Vaccine, 20 Million PFU Vaccine, 100 Million PFU Placebo Overall
Characteristic (N = 20) (N = 20) (N = 20) (N = 18) (N = 78)
Sex — no. (%)
Male 13 (65) 16 (80) 15 (75) 11 (61) 55 (71)
Female 7 (35) 4 (20) 5 (25) 7 (39) 23 (29)
Age — yr 36.9 ±11.8 34.6±12.2 38.8±14.7 32.7±10.7 35.8±12.4
Race — no. (%)†
Asian 4 (20) 3 (15) 1 (5) 2 (11) 10 (13)
Black 4 (20) 7 (35) 8 (40) 4 (22) 23 (29)
White 10 (50) 10 (50) 10 (50) 12 (67) 42 (54)
Multiracial 2 (10) 0 1 (5) 0 3 (4)
Hispanic ethnic group 1 (5) 4 (20) 1 (5.0) 0 6 (8)
— no. (%)†
Body-mass index‡ 26.3±4.7 26.7±5.2 28.7±7.9 27±6.6 27±6.2

* Plus–minus values are means ±SD. There were no significant between-group differences at baseline. PFU denotes plaque-forming units.
† Race and ethnic group were self-reported.
‡ The body-mass index is the weight in kilograms divided by the square of the height in meters.

minimum and maximum values. A two-sample assigned to receive saline placebo. At the NIH
t-test was performed for comparison of geomet- site, all the participants received a second identi-
ric mean titers between dose levels and study cal dose of vaccine 28 days after the initial dose.
sites. A paired t-test was used for comparisons All the volunteers completed the follow-up visits
between time points within a dose level. All that were scheduled during the 28-day windows
calculations and comparisons of geometric mean after vaccination; however, 4 volunteers were lost
titers were performed on the log10 scale. to follow-up by the conclusion of the trial (Fig. S1
A positive response for the Kikwit strain in the Supplementary Appendix). Additional de-
ELISA was defined as a titer of 50 or more, with tails regarding the demographic characteristics
titers of less than 50 assigned values of 25 for of the volunteers are provided in Table 1.
calculation. A positive response for the PsVNA
was defined as a titer of 20 or more, with titers Safety
of less than 20 assigned values of 10 for calcula- There were no deaths, serious adverse events, or
tion. Seroconversion on these assays was de- adverse events resulting in withdrawal from the
fined as a quadrupling of the titer over the study. There was no association between vaccine
baseline value. Baseline values were subtracted dose and the frequency or severity of adverse
from the postvaccination values for determina- events (Fig. 1, and Figs. S2 and S3 and Table S10
tion of the Mayinga strain ELISA titers, as de- in the Supplementary Appendix). After a single in-
scribed previously.3,4 oculation of vaccine, mild-to-moderate injection-
site pain was observed in the majority of par-
ticipants. Systemic reactogenicity was transient
R e sult s
and, in the majority of volunteers, mild to moder-
Study Participants ate in severity. Objective fever was noted in 20 of
A total of 78 volunteers (55 men [71%] and 23 the 60 vaccinees: 11 (18%) had grade 1 fever
women [29%]), with a mean age of 36 years (temperature range, 38.0 to 38.4°C), 7 (12%) had
(range, 20 to 64), were enrolled in a consecutive grade 2 fever (temperature range, 38.5 to 38.9°C),
manner; injections were administered between and 2 (3%) had grade 3 fever (temperature range,
October 10, 2014, and January 6, 2015. A total of 39.0 to 40.0°C). Fever onset and frequency did
60 volunteers were randomly assigned to receive not appear to be dose-dependent (Fig. 1, and Fig.
rVSV-ZEBOV, and 18 volunteers were randomly S2 in the Supplementary Appendix); fever typically

n engl j med 376;4 nejm.org  January 26, 2017 333


The New England Journal of Medicine
Downloaded from nejm.org on August 28, 2023. For personal use only. No other uses without permission.
Copyright © 2017 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

Grade 1 Grade 2 Grade 3


Adverse Event
Cohort 1
Cohort 2
Abdominal Pain
Cohort 3
Placebo
Cohort 1
Cohort 2
Arthralgia
Cohort 3
Placebo
Cohort 1
Cohort 2
Chills
Cohort 3
Placebo
Cohort 1
Cohort 2
Diaphoresis
Cohort 3
Placebo
Cohort 1
Cohort 2
Diarrhea
Cohort 3
Placebo
Cohort 1
Cohort 2
Fatigue
Cohort 3
Placebo
Cohort 1
Cohort 2
Fever, Objective
Cohort 3
Placebo
Cohort 1
Cohort 2
Fever, Subjective
Cohort 3
Placebo
Cohort 1
Cohort 2
Headache
Cohort 3
Placebo
Cohort 1
Injection-Site Cohort 2
Pain Cohort 3
Placebo
Cohort 1
Injection-Site Cohort 2
Swelling Cohort 3
Placebo
Cohort 1
Cohort 2
Myalgia
Cohort 3
Placebo
Cohort 1
Cohort 2
Nausea
Cohort 3
Placebo
0 10 20 30 40 50 60 70 80 90 100
Participants (%)

334 n engl j med 376;4 nejm.org January 26, 2017

The New England Journal of Medicine


Downloaded from nejm.org on August 28, 2023. For personal use only. No other uses without permission.
Copyright © 2017 Massachusetts Medical Society. All rights reserved.
Recombinant Vesicular Stomatitis Virus Ebola Vaccine

Figure 1 (facing page). Frequency of Solicited Adverse


by day 3 after vaccination. Mild-to-moderate
Events According to Cohort and Grade. neutropenia, which occurred in 14 of 60 partici-
Cohort 1 received a dose of 3 million plaque-forming pants, was most notable on day 3 after vaccina-
units (PFU) of the vaccine, Cohort 2 a dose of 20 mil- tion and typically abated within 2 to 4 days. An
lion PFU, and Cohort 3 a dose of 100 million PFU. All asymptomatic grade 2 thrombocytopenia, asso-
adverse events were assessed for relatedness to the ciated with grade 1 lymphopenia, was noted on
vaccine; events that were judged by the investigating
physicians not to be related to the vaccine are not shown.
day 1 after vaccination in one volunteer who re-
Adverse events were graded for severity on the basis ceived a dose of 20 million PFU; the condition
of Food and Drug Administration toxicity grading.15 resolved by day 7.
Unsolicited adverse events and laboratory adverse After a report from a phase 1 study in Geneva
events are shown in the Supplementary Appendix. of the onset of arthritis in 22% of the partici-
pants starting the second week after injection,17,18
volunteers were specifically queried about the
developed 12 to 24 hours after vaccination and development of new arthralgia, arthritis, or rash
resolved by the end of postvaccination day 1. during the second week or later after vaccina-
One volunteer who received a dose of 3 million tion. A total of 19 participants reported arthral-
PFU had grade 1 fever 7 days after vaccination gia, typically soon after vaccination. Five partici-
that resolved within 24 hours without develop- pants had an onset of arthralgia 7 to 14 days
ment of other symptoms. after vaccination, and 3 participants had arthral-
Other commonly reported systemic symptoms gia that began after the second vaccination. No
among vaccinees were headache, myalgia, and clinical cases of arthritis were diagnosed.
fatigue, with typical onset 12 to 24 hours after
vaccination. Notable adverse events were unilat- rVSV-ZEBOV on PCR Assay
eral conjunctivitis that developed in one volun- PCR results are shown in Table 2. All the vacci-
teer 1 day after inoculation and oral ulcers that nated volunteers had detectable vaccine viremia
developed in five vaccinated volunteers 4 to 16 days at the first visit after vaccination (day 1 at the
after vaccination. PCR analysis of swabs of the WRAIR and day 3 at the NIH). Twelve of the 60
affected areas (a conjunctival swab and swabs of vaccinated volunteers (20%) had viremia on day 7
three of the five oral ulcers) was negative for the after vaccination. Viremia was undetectable by
Ebola glycoprotein gene insert. Three vaccinated day 14 in all vaccinees tested at that time point
participants had cervical lymphadenopathy; one (30 volunteers at the WRAIR). In the group that
of the three also reported an oral ulcer. Infec- received a dose of 3 million PFU, there was one
tious colitis developed in one participant 21 days positive urine sample on day 3 and one on day 7.
after vaccination; symptoms included severe ab- Across the vaccine groups, a small number of
dominal pain on the left side and four episodes saliva samples were PCR-positive on days 1, 3, 7,
of mild diarrhea with blood. Computed tomog- and 14. Two subsequent saliva samples were
raphy of the abdomen at an outside hospital PCR-negative in the single volunteer who had a
showed mild thickening of the descending colon. positive PCR saliva sample on day 14. Cycle-
All conditions resolved without complications. threshold values for the positive urine sample on
Among participants who received a second dose day 7 and saliva sample on day 14 were near the
of the vaccine, reactogenicity at the injection site lower limit of detection for the assay.
and systemic reactogenicity were less severe after After administration of a second vaccine dose
the second dose than after the first dose. A com- at the NIH site, a single volunteer in the group
plete list of solicited and unsolicited adverse that received a dose of 100 million PFU had vi-
events is provided in Table S10 in the Supple- remia 3 days later. PCR results were otherwise
mentary Appendix. negative in blood, urine, and saliva.
Safety laboratory values were generally unre-
markable, with the majority of adverse events ELISA for Ebola Glycoprotein
occurring after the first dose of vaccine. Transient ELISA results are shown in Figure 2 and Table 3,
mild-to-moderate lymphopenia occurred in 24 of and Tables S1 through S4 in the Supplementary
60 participants, typically on day 1, with abatement Appendix. After a single dose of vaccine, IgG

n engl j med 376;4 nejm.org  January 26, 2017 335


The New England Journal of Medicine
Downloaded from nejm.org on August 28, 2023. For personal use only. No other uses without permission.
Copyright © 2017 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

Table 2. Vaccine Virus Detection by Means of Qualitative Reverse-Transcriptase–Polymerase-Chain-Reaction Assay.

Type of Specimen Day 1* Day 3 Day 7 Day 14* Day 31† Day 35†

no. of positive samples/no. of samples tested (%)


Blood
Vaccine, 3 million PFU 10/10 (100) 20/20 (100) 1/20 (5) 0/10 0/10 0/10
Vaccine, 20 million PFU 10/10 (100) 20/20 (100) 5/20 (25) 0/10 0/9 0/10
Vaccine, 100 million PFU 10/10 (100) 19/20 (95) 6/20 (30) 0/10 1/8 (12) 0/9
Urine
Vaccine, 3 million PFU 0/10 1/10 (10) 1/10 (10) 0/10 0/10 0/10
Vaccine, 20 million PFU 0/10 0/17 0/20 0/10 0/9 0/10
Vaccine, 100 million PFU 0/10 0/19 0/19 0/10 0/8 0/9
Saliva
Vaccine, 3 million PFU 0/10 2/20 (10) 0/20 0/10 0/10 0/10
Vaccine, 20 million PFU 0/10 0/19 5/20 (25) 1/10 (10) 0/10 0/10
Vaccine, 100 million PFU 1/10 (10) 0/19 1/19 (5) 0/10 0/9 0/9

* Data for days 1 and 14 are from the Walter Reed Army Institute of Research only.
† Data for days 31 and 35 (after the second dose) are from the National Institutes of Health (NIH) Clinical Center only.

responses were observed. A total of 16 of 20 4079 in the group receiving a dose of 100 mil-
volunteers (80%) who received a dose of 3 mil- lion PFU (P = 0.01)). There was no significant dif-
lion PFU, 19 of 20 volunteers (95%) who received ference in the geometric mean titer between the
a dose of 20 million PFU, and 18 of 20 who re- group that received a dose of 20 million PFU and
ceived a dose of 100 million PFU had undergone the group that received a dose of 100 million PFU.
seroconversion by day 14. All 60 vaccinated vol- At day 28, there were no significant differ-
unteers (100%) had undergone seroconversion by ences in the geometric mean titer between the
day 28. The groups that received a dose of 20 mil- groups that were to receive a second vaccine
lion PFU or 100 million PFU had higher geomet- dose and those that were not. All three groups
ric titers against the Zaire–Kikwit strain than the that received a second dose had increases in titers
group that received a dose of 3 million PFU, both from day 28 to day 56; titers increased from
on day 14 (857 and 888 vs. 283; P = 0.008 and 1300 on day 28 to 4222 on day 56 in the group
P = 0.02, respectively) and on day 28 (4079 and that received a dose of 3 million PFU (P<0.001),
4079 vs. 1300; P = 0.001 and P<0.001, respective- from 5198 to 7352 in the group that received a
ly). All vaccinated cohorts showed increases in dose of 20 million PFU (P = 0.27), and from 3676
titers from day 14 to day 28; titers increased to 11,143 in the group that received a dose of
from 283 on day 14 to 1300 on day 28 in the 100 million PFU (P<0.001). Among participants
group receiving a dose of 3 million PFU (P<0.001), who received a second dose, the geometric mean
from 857 to 4079 in the group receiving a dose titer was higher at day 84 than at day 28 in the
of 20 million PFU (P<0.001), and from 888 to group that received a dose of 3 million PFU

Figure 2 (facing page). Antibody Responses to Ebola Glycoprotein.


Individual antibody titers as assessed at 14 and 28 days after vaccination are shown according to vaccine dose
group, as measured by an enzyme-linked immunosorbent assay (ELISA) against the Zaire–Kikwit strain glycoprotein
(Panel A) and a pseudovirion neutralization assay (Panel B). Geometric mean titers (horizontal lines) are shown for
each group and time point. Geometric mean titers from 28 days after initial vaccination through 180 days after ini-
tial vaccination are shown for the glycoprotein ELISA (Panel C) and the pseudovirion neutralization assay (Panel D).
Solid lines indicate groups that received a second dose at day 28, and dashed lines indicate groups that did not re-
ceive a second dose at day 28. In all panels, I bars indicate 95% confidence intervals.

336 n engl j med 376;4 nejm.org  January 26, 2017

The New England Journal of Medicine


Downloaded from nejm.org on August 28, 2023. For personal use only. No other uses without permission.
Copyright © 2017 Massachusetts Medical Society. All rights reserved.
Recombinant Vesicular Stomatitis Virus Ebola Vaccine

A Zaire–Kikwit Glycoprotein ELISA at 14 and 28 Days B Pseudovirion Neutralization Assay at 14 and 28 Days
5.0 5.0

4.5 4.5

4.0 4.0
Log10 Geometric Mean Titer

Log10 Geometric Mean Titer


3.5 3.5

3.0 3.0

2.5 2.5

2.0 2.0

1.5 1.5

1.0 1.0

0.0 0.0
Day 14 Day 28 Day 14 Day 28 Day 14 Day 28 Day 14 Day 28 Day 14 Day 28 Day 14 Day 28
Vaccine, Vaccine, Vaccine, Vaccine, Vaccine, Vaccine,
3 Million PFU 20 Million PFU 100 Million PFU 3 Million PFU 20 Million PFU 100 Million PFU

C Zaire–Kikwit Glycoprotein ELISA from Day 28 to Day 180


4.5

4.0
Log10 Titer

3.5

Vaccine, 3 million PFU


3.0
Vaccine, 20 million PFU
Vaccine, 100 million PFU
2.5

0.0
28 56 84 180
Day

D Pseudovirion Neutralization Assay from Day 28 to Day 180


3.5
Vaccine, 3 million PFU
3.0 Vaccine, 20 million PFU
Vaccine, 100 million PFU
2.5
Log10 Titer

2.0

1.5

1.0

0.0
28 56 84 180
Day

n engl j med 376;4 nejm.org  January 26, 2017 337


The New England Journal of Medicine
Downloaded from nejm.org on August 28, 2023. For personal use only. No other uses without permission.
Copyright © 2017 Massachusetts Medical Society. All rights reserved.
338
Table 3. Geometric Mean Antibody Titers.*

Study Group Day 14† Day 28† Day 56 Day 84 Day 180

With Second Without Second With Second Without Second With Second Without Second
Vaccination‡ Vaccination Vaccination‡ Vaccination Vaccination‡ Vaccination

geometric mean titer (95% CI)


Zaire–Kikwit glycoprotein
The

ELISA
Vaccine, 3 million PFU 283 1300 4222 2599 2986 2263 3200 2786
(150–534) (831–2034) (2478–7195) (1537–4395) (1823–4889) (1485–3449) (1878–5452) (1248–6218)
Vaccine, 20 million PFU 857 4079 7352 3733 4222 2743 3676 2540
(502–1465) (2601–6396) (4972–10,871) (2085–6682) (3269–5455) (1634–4604) (2486–5435) (1196–5396)
Vaccine, 100 million PFU 888 4079 11,143 4525 7352 3940 5572 2786
(448–1760) (2740–6070) (8143–15,248) (1933–10,597) (4972–10,871) (1501–10,343) (3339–9298) (1169–6638)
Placebo§ 29 29 30 27 31
(23–38) (23–38) (25–37) (23–32) (20–47 )
PsVNA
Vaccine, 3 million PFU 39 223 344 138 33 N/A 36 26
n e w e ng l a n d j o u r na l

(24–62) (145–342) (203–583) (74–256) (15–69) (16–81) (10–71)

The New England Journal of Medicine


of

Vaccine, 20 million PFU 47 441 653 170 47 N/A 35 23


(20–107) (236–825) (468–911) (106–275) (28–81) (17–70) (10–52)
Vaccine, 100 million PFU 127 461 669 219 90 N/A 47 46

n engl j med 376;4 nejm.org  January 26, 2017


(67–242) (312–681) (418–1071) (98–486) (47–173) (16–141) (21–103)

Copyright © 2017 Massachusetts Medical Society. All rights reserved.


m e dic i n e

Placebo§ 10 10 10 10 10
(10-–10) (10–10) (10–10) (10–10) (10–10)

* At day 28, geometric mean titers of antibodies against ZEBOV glycoprotein were higher in the groups that received a vaccine dose of 20 million PFU or 100 million PFU than in the
group that received a dose of 3 million PFU, both as assessed by an enzyme-linked immunosorbent assay (ELISA) (4079 and 4079 vs. 1300; P = 0.001 and <0.001, respectively) and as
assessed by a pseudovirion neutralization assay (PsVNA) (441 and 461 vs. 223; P = 0.07 and P = 0.01, respectively). For the PsVNA, the day 84 analysis was performed only at the NIH

Downloaded from nejm.org on August 28, 2023. For personal use only. No other uses without permission.
Clinical Center. CI denotes confidence interval, and NA not applicable.
† Analyses at day 14 and day 28 include data from volunteers at both study sites (all volunteers with only one vaccination at these time points).
‡ The second vaccination was administered on day 28.
§ Volunteers in the placebo group received no vaccinations.
Recombinant Vesicular Stomatitis Virus Ebola Vaccine

(1300 at day 28 vs. 2986 at day 84 [P = 0.02]) and [P = 0.33], and 476 vs. 669 in the group that re-
in the group that received a dose of 100 PFU ceived a dose of 100 million PFU [P = 0.19]).
(3676 at day 28 vs. 7352 at day 84 [P = 0.02]). However, this trend was reversed in titers mea-
Among participants who did not receive a sec- sured 2 months after revaccination (222 at day
ond dose, only the group that received a dose of 28 vs. 33 at day 84 in the group that received a
3 million PFU had significant increases in the geo­ dose of 3 million PFU [P<0.001], 415 vs. 47 in
metric mean titer from day 28 through day 84 the group that received a dose of 20 million PFU
(1300 at day 28 vs. 2599 at day 56 [P<0.001] and [P = 0.003], and 476 vs. 90 in the group that re-
1300 at day 28 vs. 2263 at day 84 [P = 0.003]). In ceived a dose of 100 million PFU [P<0.001]).
all three vaccinated groups, participants who re- Vaccine groups that did not receive a second
ceived a second vaccination had higher geomet- dose had a decrease in neutralizing antibody
ric mean titers on day 56 than those who did not responses from day 28 to 56 (223 vs. 138 in the
(4222 vs. 2599 in the group that received a dose group that received a dose of 3 million PFU
of 3 million PFU [P = 0.16], 7352 vs. 3733 in the [P = 0.06], 468 vs. 170 in the group that received
group that received a dose of 20 million PFU a dose of 20 million PFU [P = 0.008], and 447 vs.
[P = 0.04], and 11,143 vs. 4525 in the group that 219 in the group that received a dose of 100 mil-
received a dose of 100 million PFU [P = 0.04]). At lion PFU [P = 0.02]). At the 180-day follow-up,
the 180-day follow-up, there was no significant there was no significant difference in neutraliz-
difference in geometric mean titers between the ing antibody responses between the groups that
groups that received a second dose of vaccine received a second dose of vaccine and the groups
and the groups that received a single dose. that received a single dose.

PsVNA Titers
Discussion
Results with respect to neutralizing antibody ti-
ters against the Zaire–Kikwit strain glycoprotein Both a single and a second administration of the
are shown in Figure 2 and Table 3, and Table S5 rVSV-ZEBOV Ebola vaccine candidate elicited an
through S8 in the Supplementary Appendix. Af- antibody response without any safety concerns
ter a single vaccination, all groups had neutral- being identified. In 60 healthy adults, the vac-
izing antibodies by day 28, in a dose-dependent cine candidate had an acceptable safety profile
manner. The geometric mean titer in the group across all dose concentrations. The most common
that received a dose of 100 million PFU was sig- side effects were injection-site pain, myalgia,
nificantly higher than in the group that received fatigue, headache, subjective fever, and chills.
a dose of 3 million PFU both on day 14 (127 vs. Immunogenicity as measured by means of IgG
39 [P  = 0.004]) and on day 28 (461 vs. 223 ELISA was concordant with antibody responses
[P = 0.01]). All three dose groups had significant measured with the use of a functional (neutral-
increases in the geometric mean titer from day ization) assay, and the IgG ELISA results sug-
14 to day 28; the titer increased from 39 on day gested a dose response, especially between the
14 to 223 on day 28 in the group that received a group receiving a dose of 3 million PFU and the
dose of 3 million PFU (P<0.001), from 47 to 441 groups receiving higher doses, with little differ-
in the group that received a dose of 20 million ence between the group receiving a dose of 20
PFU (P<0.001), and from 127 to 461 in the group million PFU and the group receiving a dose of
that received a dose of 100 million PFU (P<0.001). 100 million PFU. Although transient arthralgia
At day 28, there were no significant differ- was noted in a minority of volunteers, clinical
ences in the geometric mean titer between the arthritis, which was reported in another clinical
groups that were to receive a second vaccine trial of the vaccine candidate, was not observed
dose and those that were not. Groups that re- at the WRAIR or NIH site. These data supported
ceived a second dose of vaccine had an initial selection of 20 million PFU as the dose for clini-
trend of increased geometric mean titers during cal end-point trials (PREVAIL trial, the Guinea
the month after revaccination (222 at day 28 vs. study, and STRIVE) in West Africa. In the Guinea
344 at day 56 in the group that received a dose study, this dose recently showed high protec-
of 3 million PFU [P = 0.08], 415 vs. 653 in the tive efficacy with the use of a ring vaccination
group that received a dose of 20 million PFU strategy.12

n engl j med 376;4 nejm.org  January 26, 2017 339


The New England Journal of Medicine
Downloaded from nejm.org on August 28, 2023. For personal use only. No other uses without permission.
Copyright © 2017 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

Transient rVSV viremia was detected after im- Although a two-dose regimen was associated
munization, recapitulating the experience de- with a short-term advantage with respect to the
scribed previously in nonhuman primates.19 The magnitude of the humoral response, we did not
clinical symptoms associated with this viremia observe a significant difference in the day 180
included fever and appeared to peak and then titer between the one-dose and two-dose vaccine
decrease in the 12 to 36 hours after vaccination. regimens. The vaccine candidate has already
Overall, safety laboratory values were subclinical shown efficacy in populations living in regions
and unremarkable. Moderate asymptomatic de- in which EBV is endemic, but the immunologic
clines in leukocyte subsets (e.g., lymphopenia and profiles presented here suggest that two doses
neutropenia) were noted during the first 3 days of vaccine administered within a short time
after vaccination and resolved rapidly. The data frame may provide increased short-term benefit.5
from the clinical trials presented herein are In addition, however, strategies such as longer
consistent with the preclinical experience and, intervals between doses could be pursued to
combined with the established attenuation of the improve the longer-term immunologic profile.
vaccine vector, provide further support for the Such work would need to go hand-in-hand with
safety of rVSV vectors.2,13,20 assessment of efficacy in the animal model and
The immunoprotective profile that is required validation of the presented immune correlates.
for the prevention of EVD remains largely un- The results reported here support the safety,
known, and mechanistic correlates of protection acceptable side-effect profile, and immunoge-
remain undefined. Successful protection in the nicity of up to two doses of the rVSV-ZEBOV
nonhuman primate model has been shown with vaccine and encourage further investigation of
various vaccine candidates, with imputation of this vaccine candidate. Most promising are the
both cellular and humoral immune responses as robust immune responses after a single dose of
correlates of protection.21,22 In the nonhuman pri- the vaccine and the rapid onset of immunity,
mate challenge model, antibody response, prin- which could be particularly useful in outbreak
cipally IgG, has been the strongest immune interventions. Although we found short-term in-
correlate of protection associated with the rVSV- creases in humoral immunity after a second dose
ZEBOV vaccine candidate.10,23,24 Although the at the 1-month interval, it remains unknown
Kikwit strain ELISA has become the primary whether this regimen will translate to improved
readout, examination of the Mayinga-strain gly- clinical efficacy. Strategies to better understand
coprotein titers (Table S9 in the Supplementary and improve immunogenicity, including assess-
Appendix) suggests that the rVSV-ZEBOV vaccine ment of dose and regimen alterations, a longer
candidate produces cross-strain glycoprotein- duration of follow-up, and cross-strain protection
specific antibodies similar to those described for against other Ebola viruses, could be pursued.
the chimpanzee adenovirus 3 vaccine candidate.3 The views expressed are those of the authors and should not be
Neutralizing antibody assays typically have been construed as official or representing the positions of the Depart-
difficult to correlate with outcomes in studies in ments of the Army, Navy, or Defense or the National Institutes of
Health (NIH). The content of this article does not necessarily re-
animals involving EVD, but the functional assay flect the views or policies of the Department of Health and Human
used for the reported trials showed a strong as- Services, nor does mention of trade names, commercial products,
sociation with protection of nonhuman primates or organizations imply endorsement by the U.S. Government.
Supported by the Intramural Research Programs of the Na-
across multiple vaccine platforms and warrants tional Institute of Allergy and Infectious Diseases, NIH; the Na-
further investigation as a correlate of protec- tional Cancer Institute, NIH (contract no. HHSN261200800001E);
tion.24-27 the Defense Threat Reduction Agency; and the Joint Vaccine Ac-
quisition Program.
A second dose of vaccine was less reactogenic Disclosure forms provided by the authors are available with
and induced less viremia than the primary dose. the full text of this article at NEJM.org.

Appendix
The authors’ full names and academic degrees are as follows: Jason A. Regules, M.D., John H. Beigel, M.D., Kristopher M. Paolino,
M.D., Jocelyn Voell, R.N., M.S., Amy R. Castellano, L.P.N., Zonghui Hu, Ph.D., Paula Muñoz, B.S., James E. Moon, M.D., Richard C.
Ruck, M.D., Jason W. Bennett, M.D., Patrick S. Twomey, M.D., Ramiro L. Gutiérrez, M.D., Shon A. Remich, M.D., Holly R. Hack, M.S.,
Meagan L. Wisniewski, Ph.D., Matthew D. Josleyn, M.S., Steven A. Kwilas, Ph.D., Nicole Van Deusen, B.S., Olivier Tshiani Mbaya, M.D.,
Yan Zhou, Ph.D., Daphne A. Stanley, M.S., Wang Jing, M.S., Kirsten S. Smith, Ph.D., Meng Shi, M.A., Julie E. Ledgerwood, D.O.,
Barney S. Graham, M.D., Nancy J. Sullivan, Ph.D., Linda L. Jagodzinski, Ph.D., Sheila A. Peel, M.S.P.H., Ph.D., Judie B. Alimonti, Ph.D.,

340 n engl j med 376;4 nejm.org  January 26, 2017

The New England Journal of Medicine


Downloaded from nejm.org on August 28, 2023. For personal use only. No other uses without permission.
Copyright © 2017 Massachusetts Medical Society. All rights reserved.
Recombinant Vesicular Stomatitis Virus Ebola Vaccine

Jay W. Hooper, Ph.D., Peter M. Silvera, Ph.D., Brian K. Martin, Ph.D., Thomas P. Monath, M.D., W. Jay Ramsey, M.D., Ph.D., Charles J.
Link, M.D., H. Clifford Lane, M.D., Nelson L. Michael, M.D., Ph.D., Richard T. Davey, Jr., M.D., and Stephen J. Thomas, M.D.
The authors’ affiliations are as follows: the Walter Reed Army Institute of Research (J.A.R., K.M.P., A.R.C., J.E.M., R.C.R., J.W.B.,
P.S.T., S.A.R., H.R.H., M.S., L.L.J., S.A.P., N.L.M., S.J.T.) and Naval Medical Research Center (R.L.G.), Silver Spring, Leidos Biomedical
Research, Frederick National Laboratory for Cancer Research (J.H.B., W.J.), and the U.S. Army Medical Research Institute of Infectious
Diseases (M.L.W., M.D.J., S.A.K., N.V.D., K.S.S., J.W.H., P.M.S.), Frederick, and the National Institute of Allergy and Infectious Dis-
eases (NIAID) (J.V., Z.H., P.M., H.C.L., R.T.D.) and NIAID Vaccine Research Center (O.T.M., Y.Z., D.A.S., J.E.L., B.S.G., N.J.S.),
Bethesda — all in Maryland; the Public Health Agency of Canada, Ottawa (J.B.A.); and BioProtection Systems–NewLink Genetics, Ames,
IA (B.K.M., T.P.M., W.J.R., C.J.L.).

References
1. Ebola situation report — 30 March macaques with the VSVDeltaG/ZEBOVGP based vaccines for prophylaxis and treat-
2016. Geneva:​World Health Organization vaccine stimulates strong Ebola GP-spe- ment of filovirus infections. J Bioterr Bio-
(http://apps​.who​.int/​ebola/​current-situation/​ cific immune responses. PLoS One 2009;​ def 2011. Special Issue 1 (http://www​
ebola-situation-report-30-march-2016). 4(5):​e5547. .omicsonline​.org/​2157-2526/​2157-2526-S1
2. Marzi A, Feldmann H. Ebola virus vac- 12. Henao-Restrepo AM, Camacho A, -004​.digital/​2157-2526-S1-004​.html).
cines: an overview of current approaches. Longini IM, et al. Efficacy and effective- 20. Mire CE, Miller AD, Carville A, et al.
Expert Rev Vaccines 2014;​13:​521-31. ness of an rVSV-vectored vaccine in pre- Recombinant vesicular stomatitis virus
3. Ledgerwood JE, DeZure AD, Stanley venting Ebola virus disease: final results vaccine vectors expressing filovirus glyco-
DA, et al. Chimpanzee adenovirus vector from the Guinea ring vaccination, open- proteins lack neurovirulence in nonhuman
Ebola vaccine — preliminary report. N Engl label, cluster-randomised trial (Ebola Ça primates. PLoS Negl Trop Dis 2012;​6(3):​
J Med. DOI:​10.1056/NEJMoa1410863. Suffit!). Lancet 2016 December 23 (Epub e1567.
4. Ewer K, Rampling T, Venkatraman N, ahead of print). 21. Hensley LE, Mulangu S, Asiedu C, et
et al. A monovalent chimpanzee adenovirus 13. Garbutt M, Liebscher R, Wahl-Jensen al. Demonstration of cross-protective vac-
Ebola vaccine boosted with MVA. N Engl J V, et al. Properties of replication-compe- cine immunity against an emerging patho-
Med 2016;​374:​1635-46. tent vesicular stomatitis virus vectors ex- genic Ebolavirus species. PLoS Pathog
5. Henao-Restrepo AM, Longini IM, Egger pressing glycoproteins of filoviruses and 2010;​6(5):​e1000904.
M, et al. Efficacy and effectiveness of an arenaviruses. J Virol 2004;​78:​5458-65. 22. Marzi A, Engelmann F, Feldmann F,
rVSV-vectored vaccine expressing Ebola 14. Kugelman JR, Sanchez-Lockhart M, et al. Antibodies are necessary for rVSV/
surface glycoprotein: interim results from Andersen KG, et al. Evaluation of the po- ZEBOV-GP-mediated protection against
the Guinea ring vaccination cluster-ran- tential impact of Ebola virus genomic drift lethal Ebola virus challenge in nonhuman
domised trial. Lancet 2015;​386:​857-66. on the efficacy of sequence-based candi- primates. Proc Natl Acad Sci U S A 2013;​
6. Watanabe S, Takada A, Watanabe T, date therapeutics. MBio 2015;​6:​e02227-14. 110:​1893-8.
Ito H, Kida H, Kawaoka Y. Functional im- 15. Guidance for industry:​toxicity grading 23. Wong G, Richardson JS, Pillet S, et al.
portance of the coiled-coil of the Ebola scale for healthy adult and adolescent vol- Immune parameters correlate with pro-
virus glycoprotein. J Virol 2000;​74:​10194- unteers enrolled in preventive vaccine clini- tection against Ebola virus infection in
201. cal trials. Silver Spring, MD:​Food and Drug rodents and nonhuman primates. Sci
7. Johnson JE, Nasar F, Coleman JW, et al. Administration, September 2007 (http:// Transl Med 2012;​4:​158ra146.
Neurovirulence properties of recombinant www​.fda​.gov/​BiologicsBloodVaccines/​ 24. Jones SM, Feldmann H, Ströher U, et
vesicular stomatitis virus vectors in non- GuidanceComplianceRegulatory al. Live attenuated recombinant vaccine
human primates. Virology 2007;​360:​36-49. Information/​Guidances/​Vaccines/​ protects nonhuman primates against
8. Geisbert TW, Daddario-Dicaprio KM, ucm074775​.htm). Ebola and Marburg viruses. Nat Med 2005;​
Lewis MG, et al. Vesicular stomatitis virus- 16. Sullivan NJ, Geisbert TW, Geisbert JB, 11:​786-90.
based Ebola vaccine is well-tolerated and et al. Immune protection of nonhuman 25. Hooper JW. Neutralization assays. In:​
protects immunocompromised nonhuman primates against Ebola virus with single Immunology of protection from Ebola vi-
primates. PLoS Pathog 2008;​4(11):​e1000225. low-dose adenovirus vectors encoding rus infection. NIH VideoCasting and Pod-
9. Geisbert TW, Feldmann H. Recombi- modified GPs. PLoS Med 2006;​3(6):​e177. casting (http://videocast​.nih​.gov/​launch​
nant vesicular stomatitis virus-based vac- 17. Agnandji ST, Huttner A, Zinser ME, et .asp?18779).
cines against Ebola and Marburg virus al. Phase 1 trials of rVSV Ebola vaccine in 26. Feldmann H, Jones SM, Daddario-
infections. J Infect Dis 2011;​204:​Suppl 3:​ Africa and Europe. N Engl J Med 2016;​ DiCaprio KM, et al. Effective post-expo-
S1075-S1081. 374:​1647-60. sure treatment of Ebola infection. PLoS
10. Geisbert TW, Daddario-Dicaprio KM, 18. Huttner A, Dayer JA, Yerly S, et al. The Pathog 2007;​3(1):​e2.
Geisbert JB, et al. Vesicular stomatitis virus- effect of dose on the safety and immuno- 27. Wong G, Audet J, Fernando L, et al.
based vaccines protect nonhuman primates genicity of the VSV Ebola candidate vac- Immunization with vesicular stomatitis
against aerosol challenge with Ebola and cine: a randomised double-blind, placebo- virus vaccine expressing the Ebola glyco-
Marburg viruses. Vaccine 2008;​26:​6894- controlled phase 1/2 trial. Lancet Infect protein provides sustained long-term
900. Dis 2015;​15:​1156-66. protection in rodents. Vaccine 2014;​32:​
11. Qiu X, Fernando L, Alimonti JB, et al. 19. Marzi A, Feldmann H, Geisbert TW, 5722-9.
Mucosal immunization of cynomolgus Falzarano D. Vesicular stomatitis virus- Copyright © 2017 Massachusetts Medical Society.

n engl j med 376;4 nejm.org  January 26, 2017 341


The New England Journal of Medicine
Downloaded from nejm.org on August 28, 2023. For personal use only. No other uses without permission.
Copyright © 2017 Massachusetts Medical Society. All rights reserved.

You might also like